Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Stem Cells ; 42(4): 385-401, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38206366

ABSTRACT

Pancreatic ductal progenitor cells have been proposed to contribute to adult tissue maintenance and regeneration after injury, but the identity of such ductal cells remains elusive. Here, from adult mice, we identify a near homogenous population of ductal progenitor-like clusters, with an average of 8 cells per cluster. They are a rare subpopulation, about 0.1% of the total pancreatic cells, and can be sorted using a fluorescence-activated cell sorter with the CD133highCD71lowFSCmid-high phenotype. They exhibit properties in self-renewal and tri-lineage differentiation (including endocrine-like cells) in a unique 3-dimensional colony assay system. An in vitro lineage tracing experiment, using a novel HprtDsRed/+ mouse model, demonstrates that a single cell from a cluster clonally gives rise to a colony. Droplet RNAseq analysis demonstrates that these ductal clusters express embryonic multipotent progenitor cell markers Sox9, Pdx1, and Nkx6-1, and genes involved in actin cytoskeleton regulation, inflammation responses, organ development, and cancer. Surprisingly, these ductal clusters resist prolonged trypsin digestion in vitro, preferentially survive in vivo after a severe acinar cell injury and become proliferative within 14 days post-injury. Thus, the ductal clusters are the fundamental units of progenitor-like cells in the adult murine pancreas with implications in diabetes treatment and tumorigenicity.


Subject(s)
Acinar Cells , Pancreatic Ducts , Mice , Animals , Pancreas , Stem Cells , Cell Differentiation
2.
Stem Cell Reports ; 18(3): 618-635, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36868230

ABSTRACT

Progenitor cells capable of self-renewal and differentiation in the adult human pancreas are an under-explored resource for regenerative medicine. Using micro-manipulation and three-dimensional colony assays we identify cells within the adult human exocrine pancreas that resemble progenitor cells. Exocrine tissues were dissociated into single cells and plated into a colony assay containing methylcellulose and 5% Matrigel. A subpopulation of ductal cells formed colonies containing differentiated ductal, acinar, and endocrine lineage cells, and expanded up to 300-fold with a ROCK inhibitor. When transplanted into diabetic mice, colonies pre-treated with a NOTCH inhibitor gave rise to insulin-expressing cells. Both colonies and primary human ducts contained cells that simultaneously express progenitor transcription factors SOX9, NKX6.1, and PDX1. In addition, in silico analysis identified progenitor-like cells within ductal clusters in a single-cell RNA sequencing dataset. Therefore, progenitor-like cells capable of self-renewal and tri-lineage differentiation either pre-exist in the adult human exocrine pancreas, or readily adapt in culture.


Subject(s)
Diabetes Mellitus, Experimental , Methylcellulose , Humans , Adult , Mice , Animals , Pancreas , Pancreatic Ducts , Stem Cells
3.
J Exp Med ; 219(6)2022 06 06.
Article in English | MEDLINE | ID: mdl-35522218

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is one of the deadliest cancers and is projected to soon be the second leading cause of cancer death. Median survival of PDA patients is 6-10 mo, with the majority of diagnoses occurring at later, metastatic stages that are refractory to treatment and accompanied by worsening prognoses. Glycosylation is one of the most common types of post-translational modifications. The complex landscape of glycosylation produces an extensive repertoire of glycan moieties, glycoproteins, and glycolipids, thus adding a dynamic and tunable level of intra- and intercellular signaling regulation. Aberrant glycosylation is a feature of cancer progression and influences a broad range of signaling pathways to promote disease onset and progression. However, despite being so common, the functional consequences of altered glycosylation and their potential as therapeutic targets remain poorly understood and vastly understudied in the context of PDA. In this review, the functionality of glycans as they contribute to hallmarks of PDA are highlighted as active regulators of disease onset, tumor progression, metastatic capability, therapeutic resistance, and remodeling of the tumor immune microenvironment. A deeper understanding of the functional consequences of altered glycosylation will facilitate future hypothesis-driven studies and identify novel therapeutic strategies in PDA.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Carcinoma, Pancreatic Ductal/metabolism , Glycosylation , Humans , Pancreatic Neoplasms/pathology , Polysaccharides/metabolism , Tumor Microenvironment , Pancreatic Neoplasms
4.
PLoS One ; 16(6): e0252233, 2021.
Article in English | MEDLINE | ID: mdl-34077449

ABSTRACT

Tumor angiogenesis is critical for the growth and progression of cancer. As such, angiostasis is a treatment modality for cancer with potential utility for multiple types of cancer and fewer side effects. However, clinical success of angiostatic monotherapies has been moderate, at best, causing angiostatic treatments to lose their early luster. Previous studies demonstrated compensatory mechanisms that drive tumor vascularization despite the use of angiostatic monotherapies, as well as the potential for combination angiostatic therapies to overcome these compensatory mechanisms. We screened clinically approved angiostatics to identify specific combinations that confer potent inhibition of tumor-induced angiogenesis. We used a novel modification of the ex ovo chick chorioallantoic membrane (CAM) model that combined confocal and automated analyses to quantify tumor angiogenesis induced by glioblastoma tumor onplants. This model is advantageous due to its low cost and moderate throughput capabilities, while maintaining complex in vivo cellular interactions that are difficult to replicate in vitro. After screening multiple combinations, we determined that glioblastoma-induced angiogenesis was significantly reduced using a combination of bevacizumab (Avastin®) and temsirolimus (Torisel®) at doses below those where neither monotherapy demonstrated activity. These preliminary results were verified extensively, with this combination therapy effective even at concentrations further reduced 10-fold with a CI value of 2.42E-5, demonstrating high levels of synergy. Thus, combining bevacizumab and temsirolimus has great potential to increase the efficacy of angiostatic therapy and lower required dosing for improved clinical success and reduced side effects in glioblastoma patients.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Chorioallantoic Membrane/drug effects , Drug Screening Assays, Antitumor/methods , Drug Synergism , Glioblastoma/drug therapy , Neovascularization, Pathologic/drug therapy , Animals , Bevacizumab/administration & dosage , Chickens , Chorioallantoic Membrane/pathology , Glioblastoma/blood supply , Glioblastoma/pathology , Humans , Neovascularization, Pathologic/pathology , Rats , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , Tumor Cells, Cultured
6.
J Biol Chem ; 294(45): 16634-16649, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31533988

ABSTRACT

The existence and regenerative potential of resident stem and progenitor cells in the adult pancreas are controversial topics. A question that has been only minimally addressed is the capacity of a progenitor cell to self-renew, a key attribute that defines stem cells. Previously, our laboratory has identified putative stem and progenitor cells from the adult murine pancreas. Using an ex vivo colony/organoid culture system, we demonstrated that these stem/progenitor-like cells have self-renewal and multilineage differentiation potential. We have named these cells pancreatic colony-forming units (PCFUs) because they can give rise to three-dimensional colonies. However, the molecular mechanisms by which PCFUs self-renew have remained largely unknown. Here, we tested the hypothesis that PCFU self-renewal requires GLIS family zinc finger 3 (GLIS3), a zinc-finger transcription factor important in pancreas development. Pancreata from 2- to 4-month-old mice were dissociated, sorted for CD133highCD71low ductal cells, known to be enriched for PCFUs, and virally transduced with shRNAs to knock down GLIS3 and other proteins. We then plated these cells into our colony assays and analyzed the resulting colonies for protein and gene expression. Our results revealed a previously unknown GLIS3-to-CD133-to-WNT signaling axis in which GLIS3 and CD133 act as factors necessary for maintaining WNT receptors and signaling molecules in colonies, allowing responses to WNT ligands. Additionally, we found that CD133, but not GLIS3 or WNT, is required for phosphoinositide 3-kinase (PI3K)/AKT Ser/Thr kinase (AKT)-mediated PCFU survival. Collectively, our results uncover a molecular pathway that maintains self-renewal of adult murine PCFUs.


Subject(s)
AC133 Antigen/metabolism , DNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Wnt Proteins/metabolism , AC133 Antigen/antagonists & inhibitors , AC133 Antigen/genetics , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Self Renewal , Cell Survival , Cells, Cultured , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Mice , Mice, Inbred C57BL , Pancreas/cytology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Thrombospondins/genetics , Thrombospondins/metabolism , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Wnt Proteins/genetics , beta Catenin/metabolism
7.
Methods Mol Biol ; 2029: 63-80, 2019.
Article in English | MEDLINE | ID: mdl-31273734

ABSTRACT

Obtaining, growing, and analysis of pancreatic progenitor cells. Adult stem and progenitor cells have been successfully used for cell-based therapies such as transplantation of hematopoietic stem cells for various diseases. Whether stem and progenitor cells in the adult pancreas can be identified and used for replacement therapy has been a highly controversial topic. To address this controversy, our laboratory has developed in vitro colony assays to detect and characterize individual pancreatic stem and progenitor-like cells. We found that a subpopulation of ductal cells in the adult murine pancreas has the abilities to self-renew and differentiate into multiple pancreatic lineages in three-dimensional space in methylcellulose-containing semisolid media. This protocol details the techniques used for culturing and characterizing these pancreatic stem and progenitor-like cells, which we have named pancreatic colony-forming units (PCFUs), as well as their progenies (colonies). The techniques presented here include dissociation of pancreases, sorting antibody-stained cells with a fluorescence-activated cell sorter, viral transduction of dissociated pancreatic cells, growth of PCFUs in semi-solid media, whole-mount immunostaining and Western blot analysis for proteins expressed in colonies, and kidney capsule transplantation of colonies for in vivo functional analysis.


Subject(s)
Pancreas/cytology , Stem Cells/cytology , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Colony-Forming Units Assay/methods , Fluorescence , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Methylcellulose/pharmacology , Mice , Pancreas/drug effects , Stem Cells/drug effects
8.
Stem Cells Dev ; 26(8): 599-607, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28095743

ABSTRACT

Adult pancreatic stem and progenitor cells may serve as an alternative source of insulin-secreting endocrine cells in cell replacement therapy for type 1 diabetes, but much remained unknown about these cells. We previously identified adult murine pancreatic progenitor-like cells that displayed in vitro self-renewal and tri-lineage differentiation activities in a three-dimensional colony/organoid assay containing 1% methylcellulose and 5% Matrigel. However, the presence of other undefined culture components, such as serum and conditioned medium, has prevented a complete understanding of the signals required for progenitor cell growth. Here, we have established a serum-free, conditioned medium-free colony assay with the inclusion of seven defined factors: epidermal growth factor (EGF), R-Spondin 1 (RSPO1), Noggin, nicotinamide, exendin-4, activin B, and vascular endothelial growth factor (VEGF)-A. The requirements for colony growth were characterized and we found that EGF and nicotinamide were necessary and sufficient for the colony growth and long-term self-renewal of these progenitors. However, the seven factor (7F) culture medium better induced colony size and self-renewal in long-term culture than EGF plus nicotinamide alone. Individual 3-week-old colonies grown in the 7F culture medium expressed ductal, acinar, and endocrine lineage markers, suggesting that tri-lineage differentiation of the tri-potent progenitors was occurring without genetic manipulation. A delayed inhibition of Notch signaling using small molecules in 2-week-old cultures enhanced endocrine gene expression in 3-week-old colonies. This better-defined colony assay system will enable our and other laboratories for in-depth mechanistic studies on the biology of these progenitor cells.


Subject(s)
Adult Stem Cells/cytology , Cell Differentiation , Culture Media, Conditioned/pharmacology , Epidermal Growth Factor/pharmacology , Insulin-Secreting Cells/cytology , Niacinamide/pharmacology , Pancreas/cytology , Acinar Cells/cytology , Acinar Cells/metabolism , Activins/pharmacology , Adult Stem Cells/drug effects , Adult Stem Cells/metabolism , Animals , Cell Proliferation , Cells, Cultured , Exenatide , Extracellular Matrix Proteins/pharmacology , Female , Insulin-Secreting Cells/metabolism , Male , Mice , Peptides/pharmacology , Receptors, Notch/genetics , Receptors, Notch/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Venoms/pharmacology
9.
J Vis Exp ; (112)2016 06 10.
Article in English | MEDLINE | ID: mdl-27340914

ABSTRACT

Stem and progenitor cells from the adult pancreas could be a potential source of therapeutic beta-like cells for treating patients with type 1 diabetes. However, it is still unknown whether stem and progenitor cells exist in the adult pancreas. Research strategies using cre-lox lineage-tracing in adult mice have yielded results that either support or refute the idea that beta cells can be generated from the ducts, the presumed location where adult pancreatic progenitors may reside. These in vivo cre-lox lineage-tracing methods, however, cannot answer the questions of self-renewal and multi-lineage differentiation-two criteria necessary to define a stem cell. To begin addressing this technical gap, we devised 3-dimensional colony assays for pancreatic progenitors. Soon after our initial publication, other laboratories independently developed a similar, but not identical, method called the organoid assay. Compared to the organoid assay, our method employs methylcellulose, which forms viscous solutions that allow the inclusion of extracellular matrix proteins at low concentrations. The methylcellulose-containing assays permit easier detection and analyses of progenitor cells at the single-cell level, which are critical when progenitors constitute a small sub-population, as is the case for many adult organ stem cells. Together, results from several laboratories demonstrate in vitro self-renewal and multi-lineage differentiation of pancreatic progenitor-like cells from mice. The current protocols describe two methylcellulose-based colony assays to characterize mouse pancreatic progenitors; one contains a commercial preparation of murine extracellular matrix proteins and the other an artificial extracellular matrix protein known as a laminin hydrogel. The techniques shown here are 1) dissociation of the pancreas and sorting of CD133(+)Sox9/EGFP(+) ductal cells from adult mice, 2) single cell manipulation of the sorted cells, 3) single colony analyses using microfluidic qRT-PCR and whole-mount immunostaining, and 4) dissociation of primary colonies into single-cell suspensions and re-plating into secondary colony assays to assess self-renewal or differentiation.


Subject(s)
Pancreas , Stem Cells , Aging , Animals , Cell Differentiation , Insulin-Secreting Cells , Mice
10.
Stem Cell Res ; 16(1): 40-53, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26691820

ABSTRACT

Progenitor cells in the adult pancreas are potential sources of endocrine beta cells for treating type 1 diabetes. Previously, we identified tri-potent progenitor cells in the adult (2-4month-old) murine pancreas that were capable of self-renewal and differentiation into duct, acinar, and endocrine cells in vitro. These progenitor cells were named pancreatic colony-forming units (PCFUs). However, because PCFUs are a minor population in the pancreas (~1%) they are difficult to study. To enrich PCFUs, strategies using cell-surface marker analyses and fluorescence-activated cell sorting were developed. We found that CD133(high)CD71(low) cells, but not other cell populations, enriched PCFUs by up to 30 fold compared to the unsorted cells. CD133(high)CD71(low) cells generated primary, secondary, and subsequent colonies when serially re-plated in Matrigel-containing cultures, suggesting self-renewal abilities. In the presence of a laminin hydrogel, CD133(high)CD71(low) cells gave rise to colonies that contained duct, acinar, and Insulin(+)Glucagon(+) double-hormonal endocrine cells. Colonies from the laminin hydrogel culture were implanted into diabetic mice, and five weeks later duct, acinar, and Insulin(+)Glucagon(-) cells were detected in the grafts, demonstrating tri-lineage differentiation potential of CD133(high)CD71(low) cells. These CD133(high)CD71(low) cells will enable future studies of putative adult pancreas stem cells in vivo.


Subject(s)
AC133 Antigen , Aging/physiology , Antigens, CD/metabolism , Cell Membrane/metabolism , Colony-Forming Units Assay , Multipotent Stem Cells/cytology , Pancreas/cytology , Receptors, Transferrin/metabolism , Acinar Cells/cytology , Animals , Cell Self Renewal , Glucagon/metabolism , Immunohistochemistry , Insulin/metabolism , Mice, Inbred C57BL , Multipotent Stem Cells/metabolism , Multipotent Stem Cells/ultrastructure , Pancreatic Ducts/cytology , Paraffin Embedding , Sequence Analysis, RNA , Tissue Fixation
11.
Nat Cell Biol ; 17(2): 183-94, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25621950

ABSTRACT

Reprogrammed glucose metabolism as a result of increased glycolysis and glucose uptake is a hallmark of cancer. Here we show that cancer cells can suppress glucose uptake by non-tumour cells in the premetastatic niche, by secreting vesicles that carry high levels of the miR-122 microRNA. High miR-122 levels in the circulation have been associated with metastasis in breast cancer patients, and we show that cancer-cell-secreted miR-122 facilitates metastasis by increasing nutrient availability in the premetastatic niche. Mechanistically, cancer-cell-derived miR-122 suppresses glucose uptake by niche cells in vitro and in vivo by downregulating the glycolytic enzyme pyruvate kinase. In vivo inhibition of miR-122 restores glucose uptake in distant organs, including brain and lungs, and decreases the incidence of metastasis. These results demonstrate that, by modifying glucose utilization by recipient premetastatic niche cells, cancer-derived extracellular miR-122 is able to reprogram systemic energy metabolism to facilitate disease progression.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Glucose/metabolism , MicroRNAs/metabolism , Astrocytes/metabolism , Base Sequence , Breast Neoplasms/ultrastructure , Bromodeoxyuridine/metabolism , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Exosomes/metabolism , Female , Fibroblasts/metabolism , Humans , Luciferases/metabolism , Lung/pathology , MicroRNAs/genetics , Molecular Sequence Data , Neoplasm Metastasis , Pyruvate Kinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL